Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 264
Filter
1.
Genes (Basel) ; 15(5)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38790182

ABSTRACT

INTRODUCTION: Cell-free nucleic acids (cf-NAs) represent a promising biomarker of various pathological and physiological conditions. Since its discovery in 1948, cf-NAs gained prognostic value in oncology, immunology, and other relevant fields. In peritoneal dialysis (PD), blood purification is performed by exposing the peritoneal membrane. Relevant sections: Complications of PD such as acute peritonitis and peritoneal membrane aging are often critical in PD patient management. In this review, we focused on bacterial DNA, cell-free DNA, mitochondrial DNA (mtDNA), microRNA (miRNA), and their potential uses as biomarkers for monitoring PD and its complications. For instance, the isolation of bacterial DNA in early acute peritonitis allows bacterial identification and subsequent therapy implementation. Cell-free DNA in peritoneal dialysis effluent (PDE) represents a marker of stress of the peritoneal membrane in both acute and chronic PD complications. Moreover, miRNA are promising hallmarks of peritoneal membrane remodeling and aging, even before its manifestation. In this scenario, with multiple cytokines involved, mtDNA could be considered equally meaningful to determine tissue inflammation. CONCLUSIONS: This review explores the relevance of cf-NAs in PD, demonstrating its promising role for both diagnosis and treatment. Further studies are necessary to implement the use of cf-NAs in PD clinical practice.


Subject(s)
Cell-Free Nucleic Acids , DNA, Mitochondrial , Peritoneal Dialysis , Humans , Peritoneal Dialysis/adverse effects , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/blood , DNA, Mitochondrial/genetics , Biomarkers , MicroRNAs/genetics , DNA, Bacterial/genetics , Peritonitis/genetics , Peritoneum/metabolism , Peritoneum/pathology
2.
Wiad Lek ; 77(1): 35-41, 2024.
Article in English | MEDLINE | ID: mdl-38431805

ABSTRACT

OBJECTIVE: Aim: To determine the role of TLR4 gene polymorphisms as risk factors for peritonitis severity in patients undergoing surgery for acute inflammatory diseases of the abdominal cavity. PATIENTS AND METHODS: Materials and Methods: The study included 139 patients who were operated on for acute abdominal diseases (acute appendicitis and cholecystitis, perforated gastric or duodenal ulcer, etc.). Depending on the number of points on the modified APACHE II scale, patients were divided into two groups: Group 1 - 1-3 points (63 patients, 45.3%) and Group 2 - 4 or more points (76 patients, 54.7%). Polymorphisms rs1927911, rs2149356 and rs4986790 were determined by polymerase chain reaction. RESULTS: Results: The rs1927911 polymorphism of the TLR4 gene was protective for the development of peritonitis (according to the allelic model, OR 0.48; 95% CI 0.27-0.84; p=0.015). Regression analysis revealed a reduced (p=0.015) risk of severe peritonitis in rs1927911 A/A or G/A genotype carriers (OR 0.42; 95% CI 0.21-0.84) compared with G/G genotype carriers. There was no effect on the severity of peritonitis of TLR4 polymorphisms rs2149356 and rs4986790. There was a tendency to increase the frequency of the mutant G rs4986790 allele in patients with severe peritonitis (χ2=2.17; p<0.001). The analysis of the association of TLR4 gene polymorphisms with the phenotype of patients showed that carriers of mutant homozygotes and heterozygotes in the presence of severe peritonitis were older, had a tendency to coagulopathy, higher leukocytosis and leukocyte clotting rate. CONCLUSION: Conclusions: Thus, the importance of TLR in the development of severe peritonitis was confirmed and the protective role of the rs1927911 promoter polymorphism was established.


Subject(s)
Peritonitis , Toll-Like Receptor 4 , Humans , Abdominal Cavity , Case-Control Studies , Genetic Predisposition to Disease , Peritonitis/genetics , Polymorphism, Single Nucleotide , Toll-Like Receptor 4/genetics
3.
Front Immunol ; 14: 1200939, 2023.
Article in English | MEDLINE | ID: mdl-37520526

ABSTRACT

Introduction: The recent discovery of TAK981(Subasumstat), the first-in-class selective inhibitor of SUMOylation, enables new immune treatments. TAK981 is already in clinical trials to potentiate immunotherapy in metastatic tumors and hematologic malignancies. Cancer patients have more than ten times higher risk of infections, but the effects of TAK981 in sepsis are unknown and previous studies on SUMO in infections are conflicting. Methods: We used TAK981 in two sepsis models; polymicrobial peritonitis (CLP) and LPS endotoxemia. Splenectomy was done in both models to study the role of spleen. Western blotting of SUMO-conjugated proteins in spleen lysates was done. Global SUMO1 and SUMO3 knockout mice were used to study the specific SUMO regulation of inflammation in LPS endotoxemia. Splenocytes adoptive transfer was done from SUMO knockouts to wild type mice to study the role of spleen SUMOylation in experimental sepsis. Results and discussion: Here, we report that inhibition of SUMOylation with TAK981 improved survival in mild polymicrobial peritonitis by enhancing innate immune responses and peritoneal bacterial clearance. Thus, we focused on the effects of TAK981 on the immune responses to bacterial endotoxin, showing that TAK981 enhanced early TNFα production but did not affect the resolution of inflammation. Splenectomy decreased serum TNFα levels by nearly 60% and TAK981-induced TNFα responses. In the spleen, endotoxemia induced a distinct temporal and substrate specificity for SUMO1 and SUMO2/3, and both were inhibited by TAK981. Global genetic depletion of SUMO1, but not SUMO3, enhanced TNFα production and metabolic acidosis. The transfer of SUMO1-null, but not wild-type, splenocytes into splenectomized wild-type mice exacerbated TNFα production and metabolic acidosis in endotoxemia. Conclusion: These results suggest that specific regulation of splenic SUMO1 can modulate immune and metabolic responses to bacterial infection.


Subject(s)
Endotoxemia , Peritonitis , SUMO-1 Protein , Animals , Mice , Lipopolysaccharides/toxicity , Mice, Knockout , Peritonitis/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Spleen/metabolism , Tumor Necrosis Factor-alpha , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism
4.
J Gastrointestin Liver Dis ; 32(2): 206-215, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37345611

ABSTRACT

BACKGROUND AND AIMS: Despite limited sensitivity, the gold standard for the diagnosis of malignant cells in ascites is still cytology. The aim of this prospective proof-of-principle study was to evaluate DNA methylation as a molecular tool for the differential diagnosis of benign and malignant ascites. METHODS: A cohort of 79 patients with malignant and non-malignant ascites was prospectively enrolled. Ascites was assessed by cytopathological and laboratory examination. Cell pellets obtained by centrifugation were analyzed for differences in DNA methylation of of long interspersed nuclear element-1 (LINE-1) and microRNA-137. Quantitative determination of methylation in bisulfite-converted DNA was performed by pyrosequencing. In a subsequent stage, we compared our data to previously published data in the field following systematic review of the literature. RESULTS: Methylation status of studied LINE-1 and microRNA-137 could be reliably detected in all samples. Systematic evaluation revealed reliable reproducibility with satisfactory short- and long-term stability against degradation. Ascites from patients with a malignancy had a significantly higher methylation level of microRNA-137 compared with patients without tumor disease, whereas patients with peritonitis had significantly decreased methylation of microRNA-137. In contrast, differences in the measurement of the methylation status of LINE-1 could only be detected between patients with portal hypertension and a combination of malignant and infectious ascites. Inflammatory cells reflecting peritonitis correlated to DNA methylation changes. CONCLUSIONS: Analysis of DNA methylation in ascites is technically feasible, well reproducible and may lead to identification of potential biomarkers for peritoneal carcinomatosis and other conditions. Inflammatory cells due to peritonitis may also be associated with DNA methylation changes and need to be considered in future studies. Profiling studied under standardized conditions will be needed to identify the appropriate biomarkers for differential diagnosis of ascites.


Subject(s)
MicroRNAs , Peritoneal Neoplasms , Peritonitis , Humans , Ascites/etiology , Ascites/genetics , Peritoneal Neoplasms/diagnosis , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/complications , DNA Methylation , Prospective Studies , Reproducibility of Results , Biomarkers , Peritonitis/diagnosis , Peritonitis/genetics , Peritonitis/complications , MicroRNAs/genetics
5.
Cannabis Cannabinoid Res ; 8(3): 510-526, 2023 06.
Article in English | MEDLINE | ID: mdl-35446129

ABSTRACT

Introduction: Our laboratory investigates changes in the respiratory pattern during systemic inflammation in various rodent models. The endogenous cannabinoid system (ECS) regulates cytokine production and mitigates inflammation. Inflammation not only affects cannabinoid (CB) 1 and CB2 receptor gene expression (Cnr1 and Cnr2), but also increases the predictability of the ventilatory pattern. Objectives: Our primary objective was to track ventilatory pattern variability and transcription of Cnr1 and Cnr2 mRNA, and of Il1b, Il6, and tumor necrosis factor-alpha (Tnfa) mRNAs at multiple time points in central and peripheral tissues during systemic inflammation induced by peritonitis. Methods: In male Sprague Dawley rats (n=24), we caused peritonitis by implanting a fibrin clot containing either 0 or 25×106 Escherichia coli intraperitoneally. We recorded breathing with whole-animal plethysmography at baseline and 1 h before euthanasia. We euthanized the rats at 3, 6, or 12 h after inoculation and harvested the pons, medulla, lung, and heart for gene expression analysis. Results: With peritonitis, Cnr1 mRNA more than Cnr2 mRNA was correlated to Il1b, Il6, and Tnfa mRNAs in medulla, pons, and lung and changed oppositely in the pons, medulla, and lung. These changes were associated with increased predictability of ventilatory pattern. Specifically, nonlinear complexity index correlated with increased Cnr1 mRNA in the pons and medulla, and coefficient of variation for cycle duration correlated with Cnr1 and Cnr2 mRNAs in the lung. Conclusion: The mRNAs for ECS receptors varied with time during the central and peripheral inflammatory response to peritonitis. These changes occurred in the brainstem, which contains the network that generates breathing pattern and thus, may participate in ventilatory pattern changes during systemic inflammation.


Subject(s)
Cannabinoids , Peritonitis , Rats , Male , Animals , Receptors, Cannabinoid , Rodentia/metabolism , Interleukin-6 , Rats, Sprague-Dawley , Endocannabinoids/metabolism , Peritonitis/genetics , Inflammation , RNA, Messenger/genetics
6.
Front Immunol ; 13: 772446, 2022.
Article in English | MEDLINE | ID: mdl-35154099

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is an insulinotropic peptide that signals through the GLP-1 receptor (GLP-1R). GLP-1R, therefore, plays a critical role in diabetes and cardiovascular disease. Whether GLP-1R is involved in inflammatory disease such as gout remains unclear. Macrophages are critical effector cells in the pathogenesis of gout, a common form of inflammatory arthritis caused by the deposition of uric acid in joints. The expression of GLP-1R at the protein level is controversial due to the lack of specificity of existing antibodies against GLP-1R. Using a transgenic mouse model expressing enhanced green fluorescent protein (EGFP) under the control of GLP-1R promoter, here we confirmed the expression of GLP-1R by macrophages. M2 type macrophages and Ly6C+ macrophages expressed higher levels of GLP-1R, compared to their counterparts. GLP-1R deficient macrophages displayed a reduced the migratory ability and an enhanced expression of interleukin (IL)-6, while the expression of IL-1ß was not affected. In monosodium urate (MSU) crystal-induced peritonitis, an experimental model of gout, the recruitment of macrophages, especially M2 macrophages, was significantly suppressed in GLP-1R knockout mice compared to wild-type mice. In conclusion, our data suggests that GLP-1R plays a critical role in macrophage migration in MSU-induced inflammation.


Subject(s)
Cell Movement/immunology , Gene Expression Regulation , Glucagon-Like Peptide-1 Receptor/genetics , Macrophages/immunology , Peritonitis/genetics , Peritonitis/immunology , Uric Acid/administration & dosage , Animals , Disease Models, Animal , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neutrophils/immunology , Peritonitis/chemically induced
7.
Nat Commun ; 13(1): 139, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013270

ABSTRACT

Oxylipins are potent biological mediators requiring strict control, but how they are removed en masse during infection and inflammation is unknown. Here we show that lipopolysaccharide (LPS) dynamically enhances oxylipin removal via mitochondrial ß-oxidation. Specifically, genetic or pharmacological targeting of carnitine palmitoyl transferase 1 (CPT1), a mitochondrial importer of fatty acids, reveal that many oxylipins are removed by this protein during inflammation in vitro and in vivo. Using stable isotope-tracing lipidomics, we find secretion-reuptake recycling for 12-HETE and its intermediate metabolites. Meanwhile, oxylipin ß-oxidation is uncoupled from oxidative phosphorylation, thus not contributing to energy generation. Testing for genetic control checkpoints, transcriptional interrogation of human neonatal sepsis finds upregulation of many genes involved in mitochondrial removal of long-chain fatty acyls, such as ACSL1,3,4, ACADVL, CPT1B, CPT2 and HADHB. Also, ACSL1/Acsl1 upregulation is consistently observed following the treatment of human/murine macrophages with LPS and IFN-γ. Last, dampening oxylipin levels by ß-oxidation is suggested to impact on their regulation of leukocyte functions. In summary, we propose mitochondrial ß-oxidation as a regulatory metabolic checkpoint for oxylipins during inflammation.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Lipid Metabolism/genetics , Mitochondria/drug effects , Oxylipins/metabolism , Peritonitis/genetics , Sepsis/genetics , Acyl-CoA Dehydrogenase, Long-Chain/blood , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Animals , Carnitine O-Palmitoyltransferase/blood , Carnitine O-Palmitoyltransferase/genetics , Coenzyme A Ligases/blood , Coenzyme A Ligases/genetics , Female , Gene Expression Regulation , Humans , Infant, Newborn , Interferon-gamma/pharmacology , Lipidomics/methods , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/blood , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Oxidation-Reduction , Peritonitis/blood , Peritonitis/chemically induced , Peritonitis/pathology , RAW 264.7 Cells , Sepsis/blood , Sepsis/pathology
8.
Front Immunol ; 12: 739953, 2021.
Article in English | MEDLINE | ID: mdl-34745110

ABSTRACT

Erianin (Eri) is the extract of Dendrobium chrysotoxum Lindl. The NLRP3 inflammasome is a multiprotein complex that plays key roles in a wide variety of chronic inflammation-driven human diseases. Nevertheless, little is known about the protection of Eri against NLRP3 inflammasome-related diseases. In this study, we demonstrated that Eri inhibited NLRP3 inflammasome activation in vitro and in vivo. Mechanistically, Eri directly interacted with NLRP3, leading to inhibition of NLRP3 inflammasome assembly. Eri associated with the Walker A motif in the NACHT domain and suppressed NLRP3 ATPase activity. In mouse models, Eri had therapeutic effects on peritonitis, gouty arthritis and type 2 diabetes, via NLRP3. More importantly, Eri was active ex vivo for synovial fluid cells and monocytes from patients with IAV infection and gout. Eri may serve as a potential novel therapeutic compound against NLRP3-driven diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Gouty/drug therapy , Bibenzyls/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Inflammasomes/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Peritonitis/drug therapy , Phenol/pharmacology , Animals , Arthritis, Gouty/genetics , Arthritis, Gouty/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Dogs , HEK293 Cells , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Madin Darby Canine Kidney Cells , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peritonitis/genetics , Peritonitis/metabolism , Protein Interaction Domains and Motifs , THP-1 Cells
9.
J Clin Invest ; 131(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34699386

ABSTRACT

Chronic inflammation is a hallmark of atherosclerosis and results from an imbalance between proinflammatory and proresolving signaling. The human GPR32 receptor, together with the ALX/FPR2 receptor, transduces biological actions of several proresolving mediators that stimulate resolution of inflammation. However, since no murine homologs of the human GPR32 receptor exist, comprehensive in vivo studies are lacking. Using human atherosclerotic lesions from carotid endarterectomies and creating a transgenic mouse model expressing human GPR32 on a Fpr2×ApoE double-KO background (hGPR32myc×Fpr2-/-×Apoe-/-), we investigated the role of GPR32 in atherosclerosis and self-limiting acute inflammation. GPR32 mRNA was reduced in human atherosclerotic lesions and correlated with the immune cell markers ARG1, NOS2, and FOXP3. Atherosclerotic lesions, necrotic core, and aortic inflammation were reduced in hGPR32mycTg×Fpr2-/-×Apoe-/- transgenic mice as compared with Fpr2-/-×Apoe-/- nontransgenic littermates. In a zymosan-induced peritonitis model, the hGPR32mycTg×Fpr2-/-×Apoe-/- transgenic mice had reduced inflammation at 4 hours and enhanced proresolving macrophage responses at 24 hours compared with nontransgenic littermates. The GPR32 agonist aspirin-triggered resolvin D1 (AT-RvD1) regulated leukocyte responses, including enhancing macrophage phagocytosis and intracellular signaling in hGPR32mycTg×Fpr2-/-×Apoe-/- transgenic mice, but not in Fpr2-/-×Apoe-/- nontransgenic littermates. Together, these results provide evidence that GPR32 regulates resolution of inflammation and is atheroprotective in vivo.


Subject(s)
Atherosclerosis , Macrophages/metabolism , Signal Transduction/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Disease Models, Animal , Docosahexaenoic Acids/genetics , Docosahexaenoic Acids/metabolism , Female , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , Mice, Knockout, ApoE , Peritonitis/chemically induced , Peritonitis/genetics , Peritonitis/metabolism , Phagocytosis/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
10.
Turk J Gastroenterol ; 32(7): 593-599, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34464323

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the important causes of mortality due to malignancy. Toll-like receptors (TLRs) are very important in liver pathophysiology in terms of their roles in the innate immune system, such as the regulation of inflammation, wound healing, stimulation of adaptive immune responses, promotion of epithelial regeneration, and carcinogenesis. In this study, we planned to examine the role of TLR1 (rs4833095, rs5743551) and nucleotide-binding oligomerization domain (NOD2) (rs2066844, rs2066845, rs2066847) polymorphisms in the development of HCC and their effects on the clinical presentation of HCC patients. METHODS: Our study was designed prospectively. Cirrhotic and HCC patients who were followed up in our clinic between January 2015 and September 2018 were included in the study. Sex, age, cirrhosis etiology, Child-Pugh class, and MELD scores were recorded. TLR1 and NOD2 polymorphisms were studied by the PCR method. RESULTS: HCC developed in 88 (31.4%) of the 280 patients who were followed up, either during the recruitment phase of our study or during the follow-up. The mean follow-up time of our patient group was 17.04 ± 11.72 months, and the mean follow-up time of HCC patients was 12.09 ± 10.26 months. TLR1 (rs5743551) polymorphism was associated with HCC development (P = .003). TLR1 (rs5743551) and NOD2 (rs2066844) polymorphisms were associated with the development of spontaneous bacterial peritonitis (SBP) in the HCC patient group (P = .013 and P = .021, respectively). CONCLUSION: We think that increased bacterial translocation in cirrhotic patients may contribute to HCC development by causing chronic inflammation, especially in patients with TLR 1 (rs5743551) polymorphism.


Subject(s)
Carcinoma, Hepatocellular , Liver Cirrhosis , Liver Neoplasms , Nod2 Signaling Adaptor Protein , Receptors, Pattern Recognition , Aged , Bacterial Translocation/genetics , Bacterial Translocation/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/physiopathology , Female , Humans , Inflammation/genetics , Inflammation/immunology , Liver Cirrhosis/etiology , Liver Cirrhosis/genetics , Liver Cirrhosis/immunology , Liver Cirrhosis/physiopathology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/physiopathology , Male , Middle Aged , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/immunology , Peritonitis/etiology , Peritonitis/genetics , Peritonitis/immunology , Peritonitis/microbiology , Polymorphism, Genetic , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/immunology
11.
Front Immunol ; 12: 693974, 2021.
Article in English | MEDLINE | ID: mdl-34163489

ABSTRACT

Platelets play a key role in the development, progression and resolution of the inflammatory response during sterile inflammation and infection, although the mechanism is not well understood. Here we show that platelet CLEC-2 reduces tissue inflammation by regulating inflammatory macrophage activation and trafficking from the inflamed tissues. The immune regulatory function of CLEC-2 depends on the expression of its ligand, podoplanin, upregulated on inflammatory macrophages and is independent of platelet activation and secretion. Mechanistically, platelet CLEC-2 and also recombinant CLEC-2-Fc accelerates actin rearrangement and macrophage migration by increasing the expression of podoplanin and CD44, and their interaction with the ERM proteins. During ongoing inflammation, induced by lipopolysaccharide, treatment with rCLEC-2-Fc induces the rapid emigration of peritoneal inflammatory macrophages to mesenteric lymph nodes, thus reducing the accumulation of inflammatory macrophages in the inflamed peritoneum. This is associated with a significant decrease in pro-inflammatory cytokine, TNF-α and an increase in levels of immunosuppressive, IL-10 in the peritoneum. Increased podoplanin expression and actin remodelling favour macrophage migration towards CCL21, a soluble ligand for podoplanin and chemoattractant secreted by lymph node lymphatic endothelial cells. Macrophage efflux to draining lymph nodes induces T cell priming. In conclusion, we show that platelet CLEC-2 reduces the inflammatory phenotype of macrophages and their accumulation, leading to diminished tissue inflammation. These immunomodulatory functions of CLEC-2 are a novel strategy to reduce tissue inflammation and could be therapeutically exploited through rCLEC-2-Fc, to limit the progression to chronic inflammation.


Subject(s)
Blood Platelets/metabolism , Cell Movement , Lectins, C-Type/metabolism , Macrophage Activation , Macrophages, Peritoneal/metabolism , Peritonitis/metabolism , Animals , Blood Platelets/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Inflammation Mediators/metabolism , Lectins, C-Type/genetics , Lipopolysaccharides , Macrophages, Peritoneal/immunology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/genetics , Peritonitis/immunology , Phagocytosis , Phenotype , RAW 264.7 Cells , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Front Immunol ; 12: 669920, 2021.
Article in English | MEDLINE | ID: mdl-33981315

ABSTRACT

Macrophages are highly plastic, key regulators of inflammation. Deregulation of macrophage activation can lead to excessive inflammation as seen in inflammatory disorders like atherosclerosis, obesity, multiple sclerosis and sepsis. Targeting intracellular metabolism is considered as an approach to reshape deranged macrophage activation and to dampen the progression of inflammatory disorders. ATP citrate lyase (Acly) is a key metabolic enzyme and an important regulator of macrophage activation. Using a macrophage-specific Acly-deficient mouse model, we investigated the role of Acly in macrophages during acute and chronic inflammatory disorders. First, we performed RNA sequencing to demonstrate that Acly-deficient macrophages showed hyperinflammatory gene signatures in response to acute LPS stimulation in vitro. Next, we assessed endotoxin-induced peritonitis in myeloid-specific Acly-deficient mice and show that, apart from increased splenic Il6 expression, systemic and local inflammation were not affected by Acly deficiency. Also during obesity, both chronic low-grade inflammation and whole-body metabolic homeostasis remained largely unaltered in mice with Acly-deficient myeloid cells. Lastly, we show that macrophage-specific Acly deletion did not affect the severity of experimental autoimmune encephalomyelitis (EAE), an experimental model of multiple sclerosis. These results indicate that, despite increasing inflammatory responses in vitro, macrophage Acly deficiency does not worsen acute and chronic inflammatory responses in vivo. Collectively, our results indicate that caution is warranted in prospective long-term treatments of inflammatory disorders with macrophage-specific Acly inhibitors. Together with our earlier observation that myeloid Acly deletion stabilizes atherosclerotic lesions, our findings highlight that therapeutic targeting of macrophage Acly can be beneficial in some, but not all, inflammatory disorders.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Encephalomyelitis, Autoimmune, Experimental/enzymology , Inflammation/enzymology , Macrophages/enzymology , Peritonitis/enzymology , ATP Citrate (pro-S)-Lyase/genetics , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/etiology , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/metabolism , Lipopolysaccharides , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein , Obesity/complications , Peptide Fragments , Peritonitis/chemically induced , Peritonitis/genetics , Peritonitis/immunology , Phenotype , Signal Transduction
13.
Ann Clin Lab Sci ; 51(2): 255-257, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33941566

ABSTRACT

Bacterial peritonitis is a key complication of Peritoneal Dialysis (PD) and a preventable cause of withdrawal from PD treatment. Infection generally arises from contamination with skin commensals during handling of the dialysis delivery system or from translocation of gastrointestinal organisms and more rarely from an environmental organism. Herein, we report the case of a 73-year-old admitted for PD-related peritonitis due to Roseomonas gilardii with an associated environmental exposure from a domestic plumbing issue. We describe the presentation, case, and antibiotic regimen progression from empiric therapy of ceftazidime and vancomycin IP to ciprofloxacin. We acknowledge the importance of performing laboratory sensitivities given the high antibiotic resistance of the Roseomonas genus. We offer that nephrologists should consider Roseomonas as a potential causative organism of peritonitis, especially when initial or further history reveals exposure to potentially contaminated water.


Subject(s)
Methylobacteriaceae/pathogenicity , Peritonitis/diagnosis , Peritonitis/microbiology , Aged , Ciprofloxacin/pharmacology , Humans , Male , Methylobacteriaceae/genetics , Peritoneal Dialysis/adverse effects , Peritonitis/genetics , Renal Dialysis/adverse effects
14.
Nat Commun ; 12(1): 2856, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001893

ABSTRACT

Neutrophils are implicated in multiple homeostatic and pathological processes, but whether functional diversity requires discrete neutrophil subsets is not known. Here, we apply single-cell RNA sequencing to neutrophils from normal and inflamed mouse tissues. Whereas conventional clustering yields multiple alternative organizational structures, diffusion mapping plus RNA velocity discloses a single developmental spectrum, ordered chronologically. Termed here neutrotime, this spectrum extends from immature pre-neutrophils, largely in bone marrow, to mature neutrophils predominantly in blood and spleen. The sharpest increments in neutrotime occur during the transitions from pre-neutrophils to immature neutrophils and from mature marrow neutrophils to those in blood. Human neutrophils exhibit a similar transcriptomic pattern. Neutrophils migrating into inflamed mouse lung, peritoneum and joint maintain the core mature neutrotime signature together with new transcriptional activity that varies with site and stimulus. Together, these data identify a single developmental spectrum as the dominant organizational theme of neutrophil heterogeneity.


Subject(s)
Neutrophils/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Female , Gene Ontology , Humans , Male , Mice, Inbred C57BL , Neutrophils/cytology , Peritonitis/genetics , Peritonitis/pathology , Pneumonia/genetics , Pneumonia/pathology , Spleen/cytology , Spleen/metabolism
15.
Int J Mol Sci ; 22(7)2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33808062

ABSTRACT

The liver is well recognized as a non-immunological visceral organ that is involved in various metabolic activities, nutrient storage, and detoxification. Recently, many studies have demonstrated that resident immune cells in the liver drive various immunological reactions by means of several molecular modulators. Understanding the mechanistic details of interactions between hepatic host immune cells, including Kupffer cells and lymphocytes, and various hepatic pathogens, especially viruses, bacteria, and parasites, is necessary. MicroRNAs (miRNAs), over 2600 of which have been discovered, are small, endogenous, interfering, noncoding RNAs that are predicted to regulate more than 15,000 genes by degrading specific messenger RNAs. Several recent studies have demonstrated that some miRNAs are associated with the immune response to pathogens in the liver. However, the details of the underlying mechanisms of miRNA interference in hepatic host-pathogen interactions still remain elusive. In this review, we summarize the relationship between the immunological interactions of various pathogens and hepatic resident immune cells, as well as the role of miRNAs in the maintenance of liver immunity against pathogens.


Subject(s)
Host-Pathogen Interactions/genetics , Liver/microbiology , Liver/parasitology , Liver/virology , MicroRNAs/genetics , Animals , Gene Expression Regulation , Hepatitis/genetics , Hepatitis Viruses/pathogenicity , Humans , Liver/immunology , Liver Abscess/genetics , Liver Abscess/microbiology , Pelvic Inflammatory Disease/genetics , Peritonitis/genetics , Peritonitis/microbiology
16.
Int Immunopharmacol ; 95: 107480, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33676148

ABSTRACT

Dexmedetomidine (DEX) is a highly selective α2-adrenoceptor agonist, which can regulate inflammatory responses. However, whether DEX interferes with the inflammation resolving remains unclear. Here, we reported the effects of DEX on zymosan-induced generalized inflammation in mice during resolution. Mice were administered intraperitoneally with DEX after the initiation of sepsis. The resolution interval (Ri), a vital resolution indice, decreased from twelve hours to eight hours after the administration of DEX. The induction of peritoneal pro-inflammatory interleukin [IL] - 1ß and tumour necrosis factor-α (TNF-α) appeared to be inhibited. Of interest, the anti-inflammatory transforming growth factor-ß1 (TGF-ß1) but not IL-10 levels were up-regulated at twenty-four hours in the DEX group along with 1.0 mg/mice zymosan A (ZyA) treatment. The expression levels of multiple genes related to protective immune processes and clearance functions were detected and revealed the same trends. DEX markedly increased the F4/80+Ly6G+ macrophage population. Additionally, the adequate apoptotic neutrophil clearance from injury after DEX installation could be reverse by opsonization or co-instillation of TGF-ß1 neutralizing antibody in vivo, promoting the inflammation-resolution programs. In conclusion, DEX post-treatment, via the increase of F4/80+Ly6G+ macrophages, provokes further secretion of TGF-ß1, leading to the attenuated cytokine storm and accelerated inflammation resolving.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Dexmedetomidine/therapeutic use , Macrophages/drug effects , Peritonitis/drug therapy , Transforming Growth Factor beta1/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, Differentiation/immunology , Antigens, Ly/immunology , Cytokines/genetics , Cytokines/immunology , Dexmedetomidine/pharmacology , Macrophages/immunology , Male , Mice, Inbred C57BL , Peritonitis/genetics , Peritonitis/immunology , Transforming Growth Factor beta1/genetics
17.
Nat Commun ; 12(1): 1770, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33741914

ABSTRACT

Inflammation generally leads to recruitment of monocyte-derived macrophages. What regulates the fate of these cells and to what extent they can assume the identity and function of resident macrophages is unclear. Here, we show that macrophages elicited into the peritoneal cavity during mild inflammation persist long-term but are retained in an immature transitory state of differentiation due to the presence of enduring resident macrophages. By contrast, severe inflammation results in ablation of resident macrophages and a protracted phase wherein the cavity is incapable of sustaining a resident phenotype, yet ultimately elicited cells acquire a mature resident identity. These macrophages also have transcriptionally and functionally divergent features that result from inflammation-driven alterations to the peritoneal cavity micro-environment and, to a lesser extent, effects of origin and time-of-residency. Hence, rather than being predetermined, the fate of inflammation-elicited peritoneal macrophages seems to be regulated by the environment.


Subject(s)
Cell Differentiation/genetics , Inflammation/genetics , Macrophages, Peritoneal/metabolism , Macrophages/metabolism , Peritoneal Cavity/pathology , Animals , Cells, Cultured , Cytokines/metabolism , Female , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Profiling , Inflammation/metabolism , Macrophages/cytology , Macrophages, Peritoneal/cytology , Male , Mice, Congenic , Mice, Inbred C57BL , Peritonitis/genetics , Peritonitis/metabolism
18.
Cell Death Dis ; 12(2): 135, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33542207

ABSTRACT

Despite the progress to understand inflammatory reactions, mechanisms causing their resolution remain poorly understood. Prostanoids, especially prostaglandin E2 (PGE2), are well-characterized mediators of inflammation. PGE2 is produced in an inducible manner in macrophages (Mϕ) by microsomal PGE2-synthase-1 (mPGES-1), with the notion that it also conveys pro-resolving properties. We aimed to characterize the role of mPGES-1 during resolution of acute, zymosan-induced peritonitis. Experimentally, we applied the mPGES-1 inhibitor compound III (CIII) once the inflammatory response was established and confirmed its potent PGE2-blocking efficacy. mPGES-1 inhibition resulted in an incomplete removal of neutrophils and a concomitant increase in monocytes and Mϕ during the resolution process. The mRNA-seq analysis identified enhanced C-X3-C motif receptor 1 (CX3CR1) expression in resident and infiltrating Mϕ upon mPGES-1 inhibition. Besides elevated Cx3cr1 expression, its ligand CX3CL1 was enriched in the peritoneal lavage of the mice, produced by epithelial cells upon mPGES-1 inhibition. CX3CL1 not only increased adhesion and survival of Mϕ but its neutralization also completely reversed elevated inflammatory cell numbers, thereby normalizing the cellular, peritoneal composition during resolution. Our data suggest that mPGES-1-derived PGE2 contributes to the resolution of inflammation by preventing CX3CL1-mediated retention of activated myeloid cells at sites of injury.


Subject(s)
Chemokine CX3CL1/metabolism , Dinoprostone/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Cells/metabolism , Macrophages, Peritoneal/drug effects , Peritonitis/enzymology , Prostaglandin-E Synthases/antagonists & inhibitors , Animals , Antibodies, Neutralizing/pharmacology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cell Adhesion , Cell Survival , Cells, Cultured , Chemokine CX3CL1/antagonists & inhibitors , Chemokine CX3CL1/genetics , Disease Models, Animal , Epithelial Cells/drug effects , Female , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Mice, Inbred C57BL , Neutrophil Infiltration , Peritonitis/genetics , Peritonitis/immunology , Phenotype , Prostaglandin-E Synthases/metabolism , Up-Regulation
19.
Int Immunopharmacol ; 91: 107287, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33378723

ABSTRACT

Natterin is an aerolysin-like pore-forming toxin responsible for the toxic effects of the venom of the medically significant fish Thalassophryne nattereri. Using a combination of pharmacologic and genetic loss-of-function approaches we conduct a systematic investigation of the regulatory mechanisms that control Natterin-induced neutrophilic inflammation in the peritonitis model. Our data confirmed the capacity of Natterin to induce a strong and sustained neutrophilic inflammation leading to systemic inflammatory lung infiltration and revealed overlapping regulatory paths in its control. We found that Natterin induced the extracellular release of mature IL-1ß and the sustained production of IL-33 by bronchial epithelial cells. We confirmed the dependence of both ST2/IL-33 and IL-17A/IL-17RA signaling on the local and systemic neutrophils migration, as well as the crucial role of IL-1α, caspase-1 and caspase-11 for neutrophilic inflammation. The inflammation triggered by Natterin was a gasdermin-D-dependent inflammasome process, despite the cells did not die by pyroptosis. Finally, neutrophilic inflammation was mediated by non-canonical NLRP6 and NLRC4 adaptors through ASC interaction, independent of NLRP3. Our data highlight that the inflammatory process dependent on non-canonical inflammasome activation can be a target for pharmacological intervention in accidents by T. nattereri, which does not have adequate specific therapy.


Subject(s)
Caspase 1/metabolism , Caspases, Initiator/metabolism , Fish Venoms/pharmacology , Inflammasomes/metabolism , Inflammation/immunology , Interleukin-1beta/metabolism , Lung/drug effects , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Peritonitis/chemically induced , Receptors, Cell Surface/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caspase 1/genetics , Caspases, Initiator/genetics , Female , Inflammasomes/immunology , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung/enzymology , Lung/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/enzymology , Neutrophils/immunology , Peritonitis/enzymology , Peritonitis/genetics , Peritonitis/immunology , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Signal Transduction
20.
Cells ; 11(1)2021 12 28.
Article in English | MEDLINE | ID: mdl-35011643

ABSTRACT

Tet methylcytosine dioxygenase 2 (Tet2) mediates demethylation of DNA. We here sought to determine the expression and function of Tet2 in macrophages upon exposure to lipopolysaccharide (LPS), and in the host response to LPS induced lung and peritoneal inflammation, and during Escherichia (E.) coli induced peritonitis. LPS induced Tet2 expression in mouse macrophages and human monocytes in vitro, as well as in human alveolar macrophages after bronchial instillation in vivo. Bone marrow-derived macrophages from myeloid Tet2 deficient (Tet2fl/flLysMCre) mice displayed enhanced production of IL-1ß, IL-6 and CXCL1 upon stimulation with several Toll-like receptor agonists; similar results were obtained with LPS stimulated alveolar and peritoneal macrophages. Histone deacetylation was involved in the effect of Tet2 on IL-6 production, whilst methylation at the Il6 promoter was not altered by Tet2 deficiency. Tet2fl/flLysMCre mice showed higher IL-6 and TNF levels in bronchoalveolar and peritoneal lavage fluid after intranasal and intraperitoneal LPS administration, respectively, whilst other inflammatory responses were unaltered. E. coli induced stronger production of IL-1ß and IL-6 by Tet2 deficient peritoneal macrophages but not in peritoneal lavage fluid of Tet2fl/flLysMCre mice after in vivo intraperitoneal infection. Tet2fl/flLysMCre mice displayed enhanced bacterial growth during E. coli peritonitis, which was associated with a reduced capacity of Tet2fl/flLysMCre peritoneal macrophages to inhibit the growth of E. coli in vitro. Collectively, these data suggest that Tet2 is involved in the regulation of macrophage functions triggered by LPS and during E. coli infection.


Subject(s)
DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , Escherichia coli/physiology , Inflammation/pathology , Lung/pathology , Myeloid Cells/metabolism , Peritonitis/pathology , Animals , Anti-Bacterial Agents/metabolism , Chemokine CXCL1/metabolism , DNA-Binding Proteins/deficiency , Dioxygenases/deficiency , Gene Expression Regulation , HEK293 Cells , Histone Deacetylases/metabolism , Humans , Inflammation/genetics , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Ligands , Lipopolysaccharides , Macrophages/metabolism , Mice , Models, Biological , Nod2 Signaling Adaptor Protein/metabolism , Peritonitis/genetics , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...